1
|
Zhang Y, Cheng H, Yu P, Wang S, Dong H, Lu S, Yang R, Li B, Luo J, Mao R, Zhang Z, Qi Y, Chen X, Ding J, He Z, Zhang J, Zhao T, Chen X, Lin R, Li H, Tian Y, Wu Y. High-throughput single-cell analysis reveals Omp38-specific monoclonal antibodies that protect against Acinetobacter baumannii infection. Emerg Microbes Infect 2025; 14:2437243. [PMID: 39614635 DOI: 10.1080/22221751.2024.2437243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/01/2024]
Abstract
Infections caused by Acinetobacter baumannii (A. baumannii) have emerged as a global public health concern because of high pathogenicity of this bacterium. Monoclonal antibodies (mAbs) have a lower likelihood of promoting drug resistance and offer targeted treatment, thereby reducing potential adverse effects; however, the therapeutic potential of mAbs targeting A. baumannii has not been fully characterized. In this study, mAbs against the outer membrane proteins (OMPs) of A. baumannii were isolated in a high-throughput manner. The ability of Omp38-specific mAbs to bind to A. baumannii strains from diverse sources was confirmed via enzyme-linked immunosorbent assay (ELISA). Intravenous administration of the Omp38-specific mAbs significantly improved the survival rate and reduced the bacterial load in a mouse model of lethal A. baumannii infection. Flow cytometry and ELISA confirmed that immune cell infiltration and cytokine production, respectively, decreased in a mouse model of sublethal A. baumannii infection. In addition, analysis of the Omp38-mAb C3 binding conformation revealed the potential mechanism of broad-spectrum binding activity of this mAb against A. baumannii. Taken together, these findings indicate that mAbs against Omp38 facilitate bacterial clearance from host, minimize inflammatory mediator release and reduce host damage, highlighting the potential of Omp38-specific mAbs in the clinical treatment of A. baumannii infection.
Collapse
Affiliation(s)
- Yiwei Zhang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Hao Cheng
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Peng Yu
- Chongqing International Institute for Immunology, Chongqing, People's Republic of China
| | - Shufeng Wang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Hui Dong
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Song Lu
- Chongqing International Institute for Immunology, Chongqing, People's Republic of China
| | - Ruiqi Yang
- Chongqing International Institute for Immunology, Chongqing, People's Republic of China
| | - Baiqing Li
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Jie Luo
- The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Ruihan Mao
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Zhaohui Zhang
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Yong Qi
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Xiaohua Chen
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Jinya Ding
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Zemin He
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Jingbo Zhang
- General Hospital of Central Theater Command, Wuhan, Hubei, People's Republic of China
| | - Tingting Zhao
- Chongqing International Institute for Immunology, Chongqing, People's Republic of China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, People's Republic of China
| | - Rong Lin
- Sanya People's Hospital, Sanya, People's Republic of China
| | - Haibo Li
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Yi Tian
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
- Chongqing International Institute for Immunology, Chongqing, People's Republic of China
| |
Collapse
|
2
|
Tian Z, Wang X, Chen J. On-chip dielectrophoretic single-cell manipulation. MICROSYSTEMS & NANOENGINEERING 2024; 10:117. [PMID: 39187499 PMCID: PMC11347631 DOI: 10.1038/s41378-024-00750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/07/2024] [Accepted: 07/07/2024] [Indexed: 08/28/2024]
Abstract
Bioanalysis at a single-cell level has yielded unparalleled insight into the heterogeneity of complex biological samples. Combined with Lab-on-a-Chip concepts, various simultaneous and high-frequency techniques and microfluidic platforms have led to the development of high-throughput platforms for single-cell analysis. Dielectrophoresis (DEP), an electrical approach based on the dielectric property of target cells, makes it possible to efficiently manipulate individual cells without labeling. This review focusses on the engineering designs of recent advanced microfluidic designs that utilize DEP techniques for multiple single-cell analyses. On-chip DEP is primarily effectuated by the induced dipole of dielectric particles, (i.e., cells) in a non-uniform electric field. In addition to simply capturing and releasing particles, DEP can also aid in more complex manipulations, such as rotation and moving along arbitrary predefined routes for numerous applications. Correspondingly, DEP electrodes can be designed with different patterns to achieve different geometric boundaries of the electric fields. Since many single-cell analyses require isolation and compartmentalization of individual cells, specific microstructures can also be incorporated into DEP devices. This article discusses common electrical and physical designs of single-cell DEP microfluidic devices as well as different categories of electrodes and microstructures. In addition, an up-to-date summary of achievements and challenges in current designs, together with prospects for future design direction, is provided.
Collapse
Affiliation(s)
- Zuyuan Tian
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB, T6G 1H9, Canada
| | - Xihua Wang
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB, T6G 1H9, Canada
| | - Jie Chen
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB, T6G 1H9, Canada.
- Academy for Engineering & Technology, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Yu P, Ran J, Yang R, Zhu H, Lu S, Wu Y, Zhao T, Xiong T. Rapid isolation of pan-neutralizing antibodies against Omicron variants from convalescent individuals infected with SARS-CoV-2. Front Immunol 2024; 15:1374913. [PMID: 38510237 PMCID: PMC10950932 DOI: 10.3389/fimmu.2024.1374913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/22/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction The emergence of SARS-CoV-2 Omicron subvariants has presented a significant challenge to global health, as these variants show resistance to most antibodies developed early in the pandemic. Therapeutic antibodies with potent efficacy to the Omicron variants are urgently demanded. Methods Utilizing the rapid antibody discovery platform, Berkeley Lights Beacon, we isolated two monoclonal neutralizing antibodies, 2173-A6 and 3462-A4. These antibodies were isolated from individuals who recently recovered from Omicron infections. Results Both antibodies, 2173-A6 and 3462-A4, demonstrated high affinity for the RBD and effectively neutralized pseudoviruses from various Omicron lineages, including BA.4/5, XBB.1.16, XBB.1.5, and EG.5.1. This neutralization was achieved through binding to identical or overlapping epitopes. Discussion The use of the Beacon platform enabled the rapid isolation and identification of effective neutralizing antibodies within less than 10 days. This process significantly accelerates the development of novel therapeutic antibodies, potentially reducing the time required to respond to unknown infectious diseases in the future.
Collapse
Affiliation(s)
- Peng Yu
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
| | - Jingping Ran
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Ruiqi Yang
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
| | - Hang Zhu
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
| | - Song Lu
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
| | - Yuzhang Wu
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
| | - Tingting Zhao
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Tianchen Xiong
- Antibody Research Platform, Chongqing International Institute for Immunology, Chongqing, China
| |
Collapse
|
4
|
Keck D, Ravi S, Yadav S, Martinez-Duarte R. The Effect of Different System Parameters on the Movement of Microbial Cells Using Light-Induced Dielectrophoresis. MICROMACHINES 2024; 15:342. [PMID: 38542589 PMCID: PMC10972513 DOI: 10.3390/mi15030342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 01/03/2025]
Abstract
The manipulation of single particles remains a topic of interest with many applications. Here we characterize the impact of selected parameters on the motion of single particles thanks to dielectrophoresis (DEP) induced by visible light, in a technique called Light-induced Dielectrophoresis, or LiDEP, also known as optoelectronic tweezers, optically induced DEP, and image-based DEP. Baker's yeast and Candida cells are exposed to an electric field gradient enabled by shining a photoconductive material with a specific pattern of visible light, and their response is measured in terms of the average cell velocity towards the gradient. The impact on cell velocity when varying the shape and color of the light pattern, as well as the distance from the cell to the pattern, is presented. The experimental setup featured a commercial light projector featuring digital light processing (DLP) technology but mechanically modified to accommodate a 40× microscope objective lens. The minimal resolution achieved on the light pattern was 8 µm. Experimental results show the capability for single cell manipulation and the possibility of using different shapes, colors, and distances to determine the average cell velocity.
Collapse
Affiliation(s)
| | | | | | - Rodrigo Martinez-Duarte
- Multiscale Manufacturing Laboratory, Department of Mechanical Engineering, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
5
|
Shokouhi AR, Chen Y, Yoh HZ, Brenker J, Alan T, Murayama T, Suu K, Morikawa Y, Voelcker NH, Elnathan R. Engineering Efficient CAR-T Cells via Electroactive Nanoinjection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304122. [PMID: 37434421 DOI: 10.1002/adma.202304122] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Jason Brenker
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Dynamic Micro Devices (DMD) Lab, Department of Mechanical & Aerospace Engineering, Monash University, 17 College Walk, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies ULVAC Inc., 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Roey Elnathan
- School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, VIC, 3216, Australia
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Waurn Ponds, VIC, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia
| |
Collapse
|
6
|
Shokouhi AR, Chen Y, Yoh HZ, Murayama T, Suu K, Morikawa Y, Brenker J, Alan T, Voelcker NH, Elnathan R. Electroactive nanoinjection platform for intracellular delivery and gene silencing. J Nanobiotechnology 2023; 21:273. [PMID: 37592297 PMCID: PMC10433684 DOI: 10.1186/s12951-023-02056-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Nanoinjection-the process of intracellular delivery using vertically configured nanostructures-is a physical route that efficiently negotiates the plasma membrane, with minimal perturbation and toxicity to the cells. Nanoinjection, as a physical membrane-disruption-mediated approach, overcomes challenges associated with conventional carrier-mediated approaches such as safety issues (with viral carriers), genotoxicity, limited packaging capacity, low levels of endosomal escape, and poor versatility for cell and cargo types. Yet, despite the implementation of nanoinjection tools and their assisted analogues in diverse cellular manipulations, there are still substantial challenges in harnessing these platforms to gain access into cell interiors with much greater precision without damaging the cell's intricate structure. Here, we propose a non-viral, low-voltage, and reusable electroactive nanoinjection (ENI) platform based on vertically configured conductive nanotubes (NTs) that allows for rapid influx of targeted biomolecular cargos into the intracellular environment, and for successful gene silencing. The localization of electric fields at the tight interface between conductive NTs and the cell membrane drastically lowers the voltage required for cargo delivery into the cells, from kilovolts (for bulk electroporation) to only ≤ 10 V; this enhances the fine control over membrane disruption and mitigates the problem of high cell mortality experienced by conventional electroporation. RESULTS Through both theoretical simulations and experiments, we demonstrate the capability of the ENI platform to locally perforate GPE-86 mouse fibroblast cells and efficiently inject a diverse range of membrane-impermeable biomolecules with efficacy of 62.5% (antibody), 55.5% (mRNA), and 51.8% (plasmid DNA), with minimal impact on cells' viability post nanoscale-EP (> 90%). We also show gene silencing through the delivery of siRNA that targets TRIOBP, yielding gene knockdown efficiency of 41.3%. CONCLUSIONS We anticipate that our non-viral and low-voltage ENI platform is set to offer a new safe path to intracellular delivery with broader selection of cargo and cell types, and will open opportunities for advanced ex vivo cell engineering and gene silencing.
Collapse
Affiliation(s)
- Ali-Reza Shokouhi
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Hao Zhe Yoh
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia
| | - Takahide Murayama
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Koukou Suu
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Yasuhiro Morikawa
- Institute of Semiconductor and Electronics Technologies, ULVAC Inc, 1220-1 Suyama, Susono, Shizuoka, 410-1231, Japan
| | - Jason Brenker
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Tuncay Alan
- Department of Mechanical and Aerospace Engineering, Monash University, Wellington Rd, Clayton, VIC, 3168, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- INM-Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC, 3168, Australia.
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, VIC, 3168, Australia.
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Melbourne, VIC, 3216, Australia.
- Institute for Frontier Materials, Deakin University, Geelong Waurn Ponds campus, Melbourne, VIC, 3216, Australia.
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong Waurn Ponds Campus, Melbourne, VIC, 3216, Australia.
| |
Collapse
|
7
|
Zhang S, Xu B, Elsayed M, Nan F, Liang W, Valley JK, Liu L, Huang Q, Wu MC, Wheeler AR. Optoelectronic tweezers: a versatile toolbox for nano-/micro-manipulation. Chem Soc Rev 2022; 51:9203-9242. [DOI: 10.1039/d2cs00359g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review covers the fundamentals, recent progress and state-of-the-art applications of optoelectronic tweezers technology, and demonstrates that optoelectronic tweezers technology is a versatile and powerful toolbox for nano-/micro-manipulation.
Collapse
Affiliation(s)
- Shuailong Zhang
- School of Mechatronical Engineering, Beijing Institute of Technology, Room 711, Building No 6, Science and Technology Park, 5 Zhongguancun South St, Haidian District, Beijing, 100081, China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| | - Bingrui Xu
- School of Mechatronical Engineering, Beijing Institute of Technology, Room 711, Building No 6, Science and Technology Park, 5 Zhongguancun South St, Haidian District, Beijing, 100081, China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
| | - Mohamed Elsayed
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Fan Nan
- Institute of Nanophotonics, Jinan University, Guangzhou 511443, China
| | - Wenfeng Liang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang, 110168, China
| | - Justin K. Valley
- Berkeley Lights, Inc, 5858 Horton Street #320, Emeryville, CA 94608, USA
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| | - Qiang Huang
- School of Mechatronical Engineering, Beijing Institute of Technology, Room 711, Building No 6, Science and Technology Park, 5 Zhongguancun South St, Haidian District, Beijing, 100081, China
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, China
| | - Ming C. Wu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
| | - Aaron R. Wheeler
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| |
Collapse
|
8
|
Hur J, Chung AJ. Microfluidic and Nanofluidic Intracellular Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004595. [PMID: 34096197 PMCID: PMC8336510 DOI: 10.1002/advs.202004595] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/14/2021] [Indexed: 05/05/2023]
Abstract
Innate cell function can be artificially engineered and reprogrammed by introducing biomolecules, such as DNAs, RNAs, plasmid DNAs, proteins, or nanomaterials, into the cytosol or nucleus. This process of delivering exogenous cargos into living cells is referred to as intracellular delivery. For instance, clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing begins with internalizing Cas9 protein and guide RNA into cells, and chimeric antigen receptor-T (CAR-T) cells are prepared by delivering CAR genes into T lymphocytes for cancer immunotherapies. To deliver external biomolecules into cells, tools, including viral vectors, and electroporation have been traditionally used; however, they are suboptimal for achieving high levels of intracellular delivery while preserving cell viability, phenotype, and function. Notably, as emerging solutions, microfluidic and nanofluidic approaches have shown remarkable potential for addressing this open challenge. This review provides an overview of recent advances in microfluidic and nanofluidic intracellular delivery strategies and discusses new opportunities and challenges for clinical applications. Furthermore, key considerations for future efforts to develop microfluidics- and nanofluidics-enabled next-generation intracellular delivery platforms are outlined.
Collapse
Affiliation(s)
- Jeongsoo Hur
- School of Biomedical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Aram J. Chung
- School of Biomedical EngineeringInterdisciplinary Program in Precision Public HealthKorea UniversitySeoul02841Republic of Korea
| |
Collapse
|
9
|
Kaladharan K, Kumar A, Gupta P, Illath K, Santra TS, Tseng FG. Microfluidic Based Physical Approaches towards Single-Cell Intracellular Delivery and Analysis. MICROMACHINES 2021; 12:631. [PMID: 34071732 PMCID: PMC8228766 DOI: 10.3390/mi12060631] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022]
Abstract
The ability to deliver foreign molecules into a single living cell with high transfection efficiency and high cell viability is of great interest in cell biology for applications in therapeutic development, diagnostics, and drug delivery towards personalized medicine. Various physical delivery methods have long demonstrated the ability to deliver cargo molecules directly to the cytoplasm or nucleus and the mechanisms underlying most of the approaches have been extensively investigated. However, most of these techniques are bulk approaches that are cell-specific and have low throughput delivery. In comparison to bulk measurements, single-cell measurement technologies can provide a better understanding of the interactions among molecules, organelles, cells, and the microenvironment, which can aid in the development of therapeutics and diagnostic tools. To elucidate distinct responses during cell genetic modification, methods to achieve transfection at the single-cell level are of great interest. In recent years, single-cell technologies have become increasingly robust and accessible, although limitations exist. This review article aims to cover various microfluidic-based physical methods for single-cell intracellular delivery such as electroporation, mechanoporation, microinjection, sonoporation, optoporation, magnetoporation, and thermoporation and their analysis. The mechanisms of various physical methods, their applications, limitations, and prospects are also elaborated.
Collapse
Affiliation(s)
- Kiran Kaladharan
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 300044, Taiwan; (K.K.); (A.K.)
| | - Ashish Kumar
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 300044, Taiwan; (K.K.); (A.K.)
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India; (P.G.); (K.I.)
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India; (P.G.); (K.I.)
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India; (P.G.); (K.I.)
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 300044, Taiwan; (K.K.); (A.K.)
| |
Collapse
|
10
|
Peng HY, Yang CM, Chen YP, Liu HL, Chen TC, Pijanowska DG, Chu PY, Hsieh CH, Wu MH. An integrated actuating and sensing system for light-addressable potentiometric sensor (LAPS) and light-actuated AC electroosmosis (LACE) operation. BIOMICROFLUIDICS 2021; 15:024109. [PMID: 33868536 PMCID: PMC8043754 DOI: 10.1063/5.0040910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/24/2021] [Indexed: 06/12/2023]
Abstract
To develop a lab on a chip (LOC) integrated with both sensor and actuator functions, a novel two-in-one system based on optical-driven manipulation and sensing in a microfluidics setup based on a hydrogenated amorphous silicon (a-Si:H) layer on an indium tin oxide/glass is first realized. A high-intensity discharge xenon lamp functioned as the light source, a chopper functioned as the modulated illumination for a certain frequency, and a self-designed optical path projected on the digital micromirror device controlled by the digital light processing module was established as the illumination input signal with the ability of dynamic movement of projected patterns. For light-addressable potentiometric sensor (LAPS) operation, alternating current (AC)-modulated illumination with a frequency of 800 Hz can be generated by the rotation speed of the chopper for photocurrent vs bias voltage characterization. The pH sensitivity, drift coefficient, and hysteresis width of the Si3N4 LAPS are 52.8 mV/pH, -3.2 mV/h, and 10.5 mV, respectively, which are comparable to the results from the conventional setup. With an identical two-in-one system, direct current illumination without chopper rotation and an AC bias voltage can be provided to an a-Si:H chip with a manipulation speed of 20 μm/s for magnetic beads with a diameter of 1 μm. The collection of magnetic beads by this light-actuated AC electroosmosis (LACE) operation at a frequency of 10 kHz can be easily realized. A fully customized design of an illumination path with less decay can be suggested to obtain a high efficiency of manipulation and a high signal-to-noise ratio of sensing. With this proposed setup, a potential LOC system based on LACE and LAPS is verified with the integration of a sensor and an actuator in a microfluidics setup for future point-of-care testing applications.
Collapse
Affiliation(s)
| | - Chia-Ming Yang
- Authors to whom correspondence should be addressed:. Tel.: +886-3-2118800 ext.: 5960 and . Tel.: +886-3-2118800 ext.: 3599
| | - Yu-Ping Chen
- Institute of Electro-Optical Engineering, Chang Gung University, Taoyuan City 333, Taiwan
| | - Hui-Ling Liu
- Department of Electronic Engineering, Chang Gung University, Taoyuan City 333, Taiwan
| | - Tsung-Cheng Chen
- Department of Electronic Engineering, Chang Gung University, Taoyuan City 333, Taiwan
| | - Dorota G. Pijanowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Science, IBBE PAS 02-109, Warsaw, Poland
| | - Po-Yu Chu
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Taoyuan City 333, Taiwan
| | | | - Min-Hsien Wu
- Authors to whom correspondence should be addressed:. Tel.: +886-3-2118800 ext.: 5960 and . Tel.: +886-3-2118800 ext.: 3599
| |
Collapse
|
11
|
Ghaemi A, Bagheri E, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. CRISPR-cas9 genome editing delivery systems for targeted cancer therapy. Life Sci 2020; 267:118969. [PMID: 33385410 DOI: 10.1016/j.lfs.2020.118969] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
The prokaryotic CRISPR-Cas systems could be applied as revolutionized genome editing tool in live cells of various species to modify, visualize and identify definite sequences of DNA and RNA. CRISPR-Cas could edit the genome by homology-directed repair and non-homologous end joining mechanisms. Furthermore, DNA-targeting modification by CRISPR-Cas methodology provides opportunity for diagnosis, therapy and the genetic disorders investigation. Here, we summarized delivery systems employed for CRISPR-Cas9 for genome editing. Then preclinical studies of the CRISPR-Cas9-based therapeutics will be discussed considering the associated challenges and developments in its translation to clinic for cancer therapy.
Collapse
Affiliation(s)
- Asma Ghaemi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Bagheri
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Ling SD, Geng Y, Chen A, Du Y, Xu J. Enhanced single-cell encapsulation in microfluidic devices: From droplet generation to single-cell analysis. BIOMICROFLUIDICS 2020; 14:061508. [PMID: 33381250 PMCID: PMC7758092 DOI: 10.1063/5.0018785] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/09/2020] [Indexed: 05/24/2023]
Abstract
Single-cell analysis to investigate cellular heterogeneity and cell-to-cell interactions is a crucial compartment to answer key questions in important biological mechanisms. Droplet-based microfluidics appears to be the ideal platform for such a purpose because the compartmentalization of single cells into microdroplets offers unique advantages of enhancing assay sensitivity, protecting cells against external stresses, allowing versatile and precise manipulations over tested samples, and providing a stable microenvironment for long-term cell proliferation and observation. The present Review aims to give a preliminary guidance for researchers from different backgrounds to explore the field of single-cell encapsulation and analysis. A comprehensive and introductory overview of the droplet formation mechanism, fabrication methods of microchips, and a myriad of passive and active encapsulation techniques to enhance single-cell encapsulation efficiency were presented. Meanwhile, common methods for single-cell analysis, especially for long-term cell proliferation, differentiation, and observation inside microcapsules, are briefly introduced. Finally, the major challenges faced in the field are illustrated, and potential prospects for future work are discussed.
Collapse
Affiliation(s)
- Si Da Ling
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yuhao Geng
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - An Chen
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jianhong Xu
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
13
|
Yu Y, Deng Y, Al Hasan MA, Bai Y, Li RZ, Deng S, Joshi P, Shin S, Hu A. Femtosecond laser-induced non-thermal welding for a single Cu nanowire glucose sensor. NANOSCALE ADVANCES 2020; 2:1195-1205. [PMID: 36133038 PMCID: PMC9419468 DOI: 10.1039/c9na00740g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/22/2020] [Indexed: 06/11/2023]
Abstract
Copper nanowires (CuNWs) are a key building block to facilitate carrier conduction across a broad range of nanodevices. For integration into nanoscale devices, manipulation and welding of these nanowires need to be overcome. Based on high energy density laser processing investigation, we report on innovative welding of single CuNWs to a silver film using a tightly focused laser beam combined with manipulation of CuNWs through the dielectrophoresis (DEP) method. Two types of lasers, femtosecond (FS) and continuous-wave (CW), were employed to analyze, improve, and control Cu-NW melting characteristics under high energy density irradiation. The FS laser welding of CuNWs resulted in a metallic joint with a low contact resistance suitable for functional electronic nanodevices. Computational simulations using the 1-D heat diffusion equation and finite difference method (FDM) were performed to gain an insight into metal-laser interactions for high performance welded contact development. Simulation studies on lasers established contrasting melting behavior of metal under laser irradiation. The device feasibility of CuNW based welded contacts was evaluated in terms of the electrical performance of a glucose sensor. It was possible to sense glucose concentration down to 10-6 M, demonstrating a path towards integration of CuNWs into wearable, flexible nanoelectronic devices.
Collapse
Affiliation(s)
- Yongchao Yu
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee Knoxville 1512 Middle Drive Knoxville TN 37996 USA
| | - Yangbao Deng
- All-solid-state Energy Storage Materials and Devices Key Laboratory of Hunan Province, College of Information and Electronic Engineering, Hunan City University Yiyang 413000 P. R. China
| | - Md Abdullah Al Hasan
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee Knoxville 1512 Middle Drive Knoxville TN 37996 USA
| | - Yanfeng Bai
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee Knoxville 1512 Middle Drive Knoxville TN 37996 USA
- College of Computer Science and Electronic Engineering, Hunan University Changsha 410082 P. R. China
| | - Ruo-Zhou Li
- College of Electronic and Optical Engineering & College Microelectronics, Nanjing University of Post and Telecommunications Nanjing 210023 P. R. China
| | - Shuguang Deng
- All-solid-state Energy Storage Materials and Devices Key Laboratory of Hunan Province, College of Information and Electronic Engineering, Hunan City University Yiyang 413000 P. R. China
| | - Pooran Joshi
- Oak Ridge National Laboratory 1 Bethel Valley Rd Oak Ridge TN 37830 USA
| | - Seungha Shin
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee Knoxville 1512 Middle Drive Knoxville TN 37996 USA
| | - Anming Hu
- Department of Mechanical, Aerospace and Biomedical Engineering, University of Tennessee Knoxville 1512 Middle Drive Knoxville TN 37996 USA
| |
Collapse
|
14
|
Dong Z, Jiao Y, Xie B, Hao Y, Wang P, Liu Y, Shi J, Chitrakar C, Black S, Wang YC, Lee LJ, Li M, Fan Y, Chang L. On-chip multiplexed single-cell patterning and controllable intracellular delivery. MICROSYSTEMS & NANOENGINEERING 2020; 6:2. [PMID: 34567617 PMCID: PMC8433345 DOI: 10.1038/s41378-019-0112-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/23/2019] [Accepted: 10/10/2019] [Indexed: 05/03/2023]
Abstract
Conventional electroporation approaches show limitations in the delivery of macromolecules in vitro and in vivo. These limitations include low efficiency, noticeable cell damage and nonuniform delivery of cells. Here, we present a simple 3D electroporation platform that enables massively parallel single-cell manipulation and the intracellular delivery of macromolecules and small molecules. A pyramid pit micropore array chip was fabricated based on a silicon wet-etching method. A controllable vacuum system was adopted to trap a single cell on each micropore. Using this chip, safe single-cell electroporation was performed at low voltage. Cargoes of various sizes ranging from oligonucleotides (molecular beacons, 22 bp) to plasmid DNA (CRISPR-Cas9 expression vectors, >9 kb) were delivered into targeted cells with a significantly higher transfection efficiency than that of multiple benchmark methods (e.g., commercial electroporation devices and Lipofectamine). The delivered dose of the chemotherapeutic drug could be controlled by adjusting the applied voltage. By using CRISPR-Cas9 transfection with this system, the p62 gene and CXCR7 gene were knocked out in tumor cells, which effectively inhibited their cellular activity. Overall, this vacuum-assisted micropore array platform provides a simple, efficient, high-throughput intracellular delivery method that may facilitate on-chip cell manipulation, intracellular investigation and cancer therapy.
Collapse
Affiliation(s)
- Zaizai Dong
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Yanli Jiao
- College of Agricultural and Life Sciences, University of Florida, Gainesville, FL 32611 USA
| | - Bingteng Xie
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yongcun Hao
- Ministry of Education Key Laboratory of Micro and Nano Systems for Aerospace, Northwestern Polytechnical University, 710072 Xi’an, China
| | - Pan Wang
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yuanyuan Liu
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Junfeng Shi
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Chandani Chitrakar
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Stephen Black
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| | - Yu-Chieh Wang
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - L. James Lee
- Chemical and Biomolecular Engineering Department, Ohio State University, Columbus, OH 43209 USA
| | - Mo Li
- Center for Reproductive Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Yubo Fan
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
| | - Lingqian Chang
- School of Biological Science and Medical Engineering, Beihang University, 100083 Beijing, China
- Institute of Nanotechnology for Single Cell Analysis (INSCA), Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, 100083 Beijing, China
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207 USA
| |
Collapse
|
15
|
Jorgolli M, Nevill T, Winters A, Chen I, Chong S, Lin F, Mock M, Chen C, Le K, Tan C, Jess P, Xu H, Hamburger A, Stevens J, Munro T, Wu M, Tagari P, Miranda LP. Nanoscale integration of single cell biologics discovery processes using optofluidic manipulation and monitoring. Biotechnol Bioeng 2019; 116:2393-2411. [PMID: 31112285 PMCID: PMC6771990 DOI: 10.1002/bit.27024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 12/12/2022]
Abstract
The new and rapid advancement in the complexity of biologics drug discovery has been driven by a deeper understanding of biological systems combined with innovative new therapeutic modalities, paving the way to breakthrough therapies for previously intractable diseases. These exciting times in biomedical innovation require the development of novel technologies to facilitate the sophisticated, multifaceted, high-paced workflows necessary to support modern large molecule drug discovery. A high-level aspiration is a true integration of "lab-on-a-chip" methods that vastly miniaturize cellulmical experiments could transform the speed, cost, and success of multiple workstreams in biologics development. Several microscale bioprocess technologies have been established that incrementally address these needs, yet each is inflexibly designed for a very specific process thus limiting an integrated holistic application. A more fully integrated nanoscale approach that incorporates manipulation, culture, analytics, and traceable digital record keeping of thousands of single cells in a relevant nanoenvironment would be a transformative technology capable of keeping pace with today's rapid and complex drug discovery demands. The recent advent of optical manipulation of cells using light-induced electrokinetics with micro- and nanoscale cell culture is poised to revolutionize both fundamental and applied biological research. In this review, we summarize the current state of the art for optical manipulation techniques and discuss emerging biological applications of this technology. In particular, we focus on promising prospects for drug discovery workflows, including antibody discovery, bioassay development, antibody engineering, and cell line development, which are enabled by the automation and industrialization of an integrated optoelectronic single-cell manipulation and culture platform. Continued development of such platforms will be well positioned to overcome many of the challenges currently associated with fragmented, low-throughput bioprocess workflows in biopharma and life science research.
Collapse
Affiliation(s)
| | - Tanner Nevill
- Product ApplicationsBerkeley Lights, IncEmeryvilleCalifornia
| | - Aaron Winters
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| | - Irwin Chen
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| | - Su Chong
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| | - Fen‐Fen Lin
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| | - Marissa Mock
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| | - Ching Chen
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| | - Kim Le
- Drug Substance Technologies, One Amgen Center DriveThousand OaksCalifornia
| | - Christopher Tan
- Drug Substance Technologies, One Amgen Center DriveThousand OaksCalifornia
| | - Philip Jess
- Product ApplicationsBerkeley Lights, IncEmeryvilleCalifornia
| | - Han Xu
- Drug DiscoveryA2 BiotherapeuticsWestlake VillageCalifornia
| | - Agi Hamburger
- Drug DiscoveryA2 BiotherapeuticsWestlake VillageCalifornia
| | - Jennitte Stevens
- Drug Substance Technologies, One Amgen Center DriveThousand OaksCalifornia
| | - Trent Munro
- Drug Substance Technologies, One Amgen Center DriveThousand OaksCalifornia
| | - Ming Wu
- Department of Electrical Engineering and Computer SciencesUniversity of California at BerkeleyBerkeleyCalifornia
| | - Philip Tagari
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| | - Les P. Miranda
- Amgen ResearchOne Amgen Center DriveThousand OaksCalifornia
| |
Collapse
|
16
|
Liang W, Liu L, Zhang H, Wang Y, Li WJ. Optoelectrokinetics-based microfluidic platform for bioapplications: A review of recent advances. BIOMICROFLUIDICS 2019; 13:051502. [PMID: 31558919 PMCID: PMC6748859 DOI: 10.1063/1.5116737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/05/2019] [Indexed: 05/14/2023]
Abstract
The introduction of optoelectrokinetics (OEK) into lab-on-a-chip systems has facilitated a new cutting-edge technique-the OEK-based micro/nanoscale manipulation, separation, and assembly processes-for the microfluidics community. This technique offers a variety of extraordinary advantages such as programmability, flexibility, high biocompatibility, low-cost mass production, ultralow optical power requirement, reconfigurability, rapidness, and ease of integration with other microfluidic units. This paper reviews the physical mechanisms that govern the manipulation of micro/nano-objects in microfluidic environments as well as applications related to OEK-based micro/nanoscale manipulation-applications that span from single-cell manipulation to single-molecular behavior determination. This paper wraps up with a discussion of the current challenges and future prospects for the OEK-based microfluidics technique. The conclusion is that this technique will allow more opportunities for biomedical and bioengineering researchers to improve lab-on-a-chip technologies and will have far-reaching implications for biorelated researches and applications in the future.
Collapse
Affiliation(s)
- Wenfeng Liang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, China
| | - Lianqing Liu
- Authors to whom correspondence should be addressed: and
| | - Hemin Zhang
- Department of Neurology, The People’s Hospital of Liaoning Province, Shenyang 110016, China
| | | | - Wen Jung Li
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
17
|
Winters A, McFadden K, Bergen J, Landas J, Berry KA, Gonzalez A, Salimi-Moosavi H, Murawsky CM, Tagari P, King CT. Rapid single B cell antibody discovery using nanopens and structured light. MAbs 2019; 11:1025-1035. [PMID: 31185801 PMCID: PMC6748590 DOI: 10.1080/19420862.2019.1624126] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Accelerated development of monoclonal antibody (mAb) tool reagents is an essential requirement for the successful advancement of therapeutic antibodies in today’s fast-paced and competitive drug development marketplace. Here, we describe a direct, flexible, and rapid nanofluidic optoelectronic single B lymphocyte antibody screening technique (NanOBlast) applied to the generation of anti-idiotypic reagent antibodies. Selectively enriched, antigen-experienced murine antibody secreting cells (ASCs) were harvested from spleen and lymph nodes. Subsequently, secreted mAbs from individually isolated, single ASCs were screened directly using a novel, integrated, high-content culture, and assay platform capable of manipulating living cells within microfluidic chip nanopens using structured light. Single-cell polymerase chain reaction–based molecular recovery on select anti-idiotypic ASCs followed by recombinant IgG expression and enzyme-linked immunosorbent assay (ELISA) characterization resulted in the recovery and identification of a diverse and high-affinity panel of anti-idiotypic reagent mAbs. Combinatorial ELISA screening identified both capture and detection mAbs, and enabled the development of a sensitive and highly specific ligand binding assay capable of quantifying free therapeutic IgG molecules directly from human patient serum, thereby facilitating important drug development decision-making. The ASC import, screening, and export discovery workflow on the chip was completed within 5 h, while the overall discovery workflow from immunization to recombinantly expressed IgG was completed in under 60 days.
Collapse
Affiliation(s)
- Aaron Winters
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - Karyn McFadden
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - John Bergen
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada
| | - Julius Landas
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada.,c Department of Pharmacokinetics & Drug Metabolism, University of British Columbia , Vancouver , Canada
| | - Kelly A Berry
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada
| | - Anthony Gonzalez
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - Hossein Salimi-Moosavi
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA.,c Department of Pharmacokinetics & Drug Metabolism, University of British Columbia , Vancouver , Canada
| | | | - Philip Tagari
- a Department of Therapeutic Discovery, Amgen Research , Thousand Oaks , CA , USA
| | - Chadwick T King
- b Department of Therapeutic Discovery, Amgen Research , Burnaby , Canada
| |
Collapse
|
18
|
Le K, Tan C, Gupta S, Guhan T, Barkhordarian H, Lull J, Stevens J, Munro T. A novel mammalian cell line development platform utilizing nanofluidics and optoelectro positioning technology. Biotechnol Prog 2018; 34:1438-1446. [PMID: 30009534 PMCID: PMC6585769 DOI: 10.1002/btpr.2690] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/21/2022]
Abstract
Generating a highly productive cell line is resource intensive and typically involves long timelines because of the need to screen large numbers of candidates in protein production studies. This has led to miniaturization and automation strategies to allow for reductions in resources and higher throughput. Current approaches rely on the use of standard cell culture vessels and bulky liquid handling equipment. New nanofludic technologies offer novel solutions to surpass these limits, further miniaturizing cell culture volumes (105 times smaller) by growing cells on custom nanofluidic chips. Berkeley Lights' OptoElectro Positioning technology projects light patterns to activate photoconductors that gently repel cells to manipulate single cells on nanofluidic culturing chips. Using a fully integrated technology platform (Beacon), common cell culture tasks can be programmed through software, allowing maintenance and analysis of thousands of cell lines in parallel on a single chip. Here, we describe the ability to perform key cell line development work on the Beacon platform. We demonstrate that commercial production Chinese hamster ovary cell lines can be isolated, cultured, screened, and exported at high efficiency. We compare this process head to head with a FACS-enabled microtiter plate-based workflow and demonstrate generation of comparable clonal cell lines with reduced resources. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1438-1446, 2018.
Collapse
Affiliation(s)
- Kim Le
- Drug Substance Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| | - Christopher Tan
- Drug Substance Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| | - Shivani Gupta
- Drug Substance Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| | - Trupti Guhan
- Drug Substance Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| | - Hedieh Barkhordarian
- Drug Substance Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| | - Jonathan Lull
- Drug Substance Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| | - Jennitte Stevens
- Drug Substance Technologies, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| | - Trent Munro
- Attribute Sciences, Process Development, Amgen Inc., Thousand Oaks, CA, 91320
| |
Collapse
|
19
|
Hoshino T, Yoshioka M, Wagatsuma A, Miyazako H, Mabuchi K. Pinpoint Delivery of Molecules by Using Electron Beam Addressing Virtual Cathode Display. IEEE Trans Nanobioscience 2018; 17:62-69. [PMID: 29570076 DOI: 10.1109/tnb.2018.2798582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Electroporation, a physical transfection method to introduce genomic molecules in selective living cells, could be implemented by microelectrode devices. A local electric field generated by a finer electrode can induces cytomembrane poration in the electrode vicinity. To employ fine, high-speed scanning electrodes, we developed a fine virtual cathode pattern, which was generated on a cell adhesive surface of 100-nm-thick SiN membrane by inverted-electron beam lithography. The SiN membrane works as both a vacuum barrier and the display screen of the virtual cathode. The kinetic energy of the incident primary electrons to the SiN membrane was completely blocked, whereas negative charges and leaking electric current appeared on the surface of the dielectric SiN membrane within a region of 100 nm. Locally controlled transmembrane molecular delivery was demonstrated on adhered C2C12 myoblast cells in a culturing medium with fluorescent dye propidium iodide (PI). Increasing fluorescence of pre-diluted PI indicated local poration and transmembrane inflow at the virtual cathode position, as well as intracellular diffusion. The transmembrane inflows depended on beam duration time and acceleration voltage. At the post-molecular delivery, a slight decrease in intracellular PI fluorescence intensity indicates membrane recovery from the poration. Cell viability was confirmed by time-lapse cell imaging of post-exposure cell migration.
Collapse
|
20
|
Li P, Yu H, Liu N, Wang F, Lee GB, Wang Y, Liu L, Li WJ. Visible light induced electropolymerization of suspended hydrogel bioscaffolds in a microfluidic chip. Biomater Sci 2018; 6:1371-1378. [PMID: 29790875 DOI: 10.1039/c7bm01153a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
3D net-like hydrogel scaffolds are fabricated via visible-light induced electropolymerization, which could be used to modulate 3D cell organization.
Collapse
Affiliation(s)
- Pan Li
- State Key Laboratory of Robotics
- Shenyang Institute of Automation
- Chinese Academy of Sciences
- Shenyang 110016
- China
| | - Haibo Yu
- State Key Laboratory of Robotics
- Shenyang Institute of Automation
- Chinese Academy of Sciences
- Shenyang 110016
- China
| | - Na Liu
- School of Mechatronics Engineering and Automation
- Shanghai University
- Shanghai 200072
- China
| | - Feifei Wang
- Department of Chemistry
- Stanford University
- Stanford
- USA
| | - Gwo-Bin Lee
- Department of Power Mechanical Engineering
- National Tsing Hua University
- Hsinchu
- Taiwan
| | - Yuechao Wang
- State Key Laboratory of Robotics
- Shenyang Institute of Automation
- Chinese Academy of Sciences
- Shenyang 110016
- China
| | - Lianqing Liu
- State Key Laboratory of Robotics
- Shenyang Institute of Automation
- Chinese Academy of Sciences
- Shenyang 110016
- China
| | - Wen Jung Li
- Department of Mechanical and Biomedical Engineering
- City University of Hong Kong
- Kowloon Tong
- Hong Kong
| |
Collapse
|
21
|
|
22
|
Controllable in-situ cell electroporation with cell positioning and impedance monitoring using micro electrode array. Sci Rep 2016; 6:31392. [PMID: 27507603 PMCID: PMC4979028 DOI: 10.1038/srep31392] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/19/2016] [Indexed: 11/22/2022] Open
Abstract
This paper reports a novel microarray chip for in-situ, real-time and selective electroporation on individual cells integrated with cell positioning and impedance monitoring. An array of quadrupole-electrode units (termed positioning electrodes) and pairs of planar center electrodes located at the centers of each quadrupole-electrode unit were fabricated on the chip. The positioning electrodes are used to trap and position living cells onto the center electrodes based on negative dielectrophoresis (nDEP). The center electrodes are used for in-situ cell electroporation, and also used to measure cell impedance for monitoring cellular dynamics in real time. Controllably selective electroporation and electrical measurement on the cells in array are realized. We present an evidence of selective electroporation through use of fluorescent dyes. Subsequently we use in-situ and real-time impedance measurement to monitor the process, which demonstrates the dynamic behavior of the cell electroporation. Finally, we show the use of this device to perform successful transfection onto individual HeLa cells with vector DNA encoding a green fluorescent.
Collapse
|
23
|
Self-Locking Optoelectronic Tweezers for Single-Cell and Microparticle Manipulation across a Large Area in High Conductivity Media. Sci Rep 2016; 6:22630. [PMID: 26940301 PMCID: PMC4778053 DOI: 10.1038/srep22630] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 02/18/2016] [Indexed: 01/23/2023] Open
Abstract
Optoelectronic tweezers (OET) has advanced within the past decade to become a promising tool for cell and microparticle manipulation. Its incompatibility with high conductivity media and limited throughput remain two major technical challenges. Here a novel manipulation concept and corresponding platform called Self-Locking Optoelectronic Tweezers (SLOT) are proposed and demonstrated to tackle these challenges concurrently. The SLOT platform comprises a periodic array of optically tunable phototransistor traps above which randomly dispersed single cells and microparticles are self-aligned to and retained without light illumination. Light beam illumination on a phototransistor turns off the trap and releases the trapped cell, which is then transported downstream via a background flow. The cell trapping and releasing functions in SLOT are decoupled, which is a unique feature that enables SLOT’s stepper-mode function to overcome the small field-of-view issue that all prior OET technologies encountered in manipulation with single-cell resolution across a large area. Massively parallel trapping of more than 100,000 microparticles has been demonstrated in high conductivity media. Even larger scale trapping and manipulation can be achieved by linearly scaling up the number of phototransistors and device area. Cells after manipulation on the SLOT platform maintain high cell viability and normal multi-day divisibility.
Collapse
|
24
|
Optically-controlled platforms for transfection and single- and sub-cellular surgery. Biophys Rev 2015; 7:379-390. [PMID: 28510103 DOI: 10.1007/s12551-015-0179-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/22/2015] [Indexed: 10/22/2022] Open
Abstract
Improving the resolution of biological research to the single-cell or sub-cellular level is of critical importance in a wide variety of processes and disease conditions. Most obvious are those linked to aging and cancer, many of which are dependent upon stochastic processes where individual, unpredictable failures or mutations in individual cells can lead to serious downstream conditions across the whole organism. The traditional tools of biochemistry struggle to observe such processes: the vast majority are based upon ensemble approaches analysing the properties of bulk populations, which means that details of individual constituents is lost. What are required, then, are tools with the precision and resolution to probe and dissect cells at the single-micron scale: the scale of the individual organelles and structures that control their function. In this review, we highlight the use of highly-focused laser beams to create systems which provide precise control and specificity at the single-cell or even single-micron level. The intense focal points generated can directly interact with cells and cell membranes, which in conjunction with related modalities such as optical trapping provide a broad platform for the development of single-cell and sub-cellular surgery approaches. These highly tuneable tools have been demonstrated to deliver or remove material from cells of interest, and they can simultaneously excite fluorescent probes for imaging purposes or plasmonic structures for very local heating. We discuss both the history and recent applications of the field, highlighting the key findings and developments over the last 40 years of biophotonics research.
Collapse
|
25
|
Fan Q, Hu W, Ohta AT. Light-induced microbubble poration of localized cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2013:4482-5. [PMID: 24110729 DOI: 10.1109/embc.2013.6610542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Molecular delivery into localized NIH/3T3 cells was achieved with microbubbles produced by laser pulses focused on an optically absorbent substrate. The laser-induced bubble expansion and contraction resulted in cell poration. The microbubbles are localized at the laser focal point, so molecular delivery can be directed at specific localized cells. This was demonstrated with the delivery of 3-kDa FITC-Dextran. Single-cell molecular delivery was achieved, even in the presence of nearby cells. The efficiency of the cell poration was up to 95%, with a corresponding cell viability of 98%.
Collapse
|
26
|
Anand RK, Johnson ES, Chiu DT. Negative dielectrophoretic capture and repulsion of single cells at a bipolar electrode: the impact of faradaic ion enrichment and depletion. J Am Chem Soc 2015; 137:776-83. [PMID: 25562315 DOI: 10.1021/ja5102689] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This paper describes the dielectrophoretic (DEP) forces generated by a bipolar electrode (BPE) in a microfluidic device and elucidates the impact of faradaic ion enrichment and depletion (FIE and FID) on electric field gradients. DEP technologies for manipulating biological cells provide several distinct advantages over other cell-handling techniques including label-free selectivity, inexpensive device components, and amenability to single-cell and array-based applications. However, extension to the array format is nontrivial, and DEP forces are notoriously short-range, limiting device dimensions and throughput. BPEs present an attractive option for DEP because of the ease with which they can be arrayed. Here, we present experimental results demonstrating both negative DEP (nDEP) attraction and repulsion of B-cells from each a BPE cathode and anode. The direction of nDEP force in each case was determined by whether the conditions for FIE or FID were chosen in the experimental design. We conclude that FIE and FID zones generated by BPEs can be exploited to shape and extend the electric field gradients that are responsible for DEP force.
Collapse
Affiliation(s)
- Robbyn K Anand
- Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | | | | |
Collapse
|
27
|
Witte C, Kremer C, Chanasakulniyom M, Reboud J, Wilson R, Cooper JM, Neale SL. Spatially selecting a single cell for lysis using light-induced electric fields. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:3026-31. [PMID: 24719234 DOI: 10.1002/smll.201400247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 02/27/2014] [Indexed: 05/16/2023]
Abstract
An optoelectronic tweezing (OET) device, within an integrated microfluidic channel, is used to precisely select single cells for lysis among dense populations. Cells to be lysed are exposed to higher electrical fields than their neighbours by illuminating a photoconductive film underneath them. Using beam spot sizes as low as 2.5 μm, 100% lysis efficiency is reached in <1 min allowing the targeted lysis of cells.
Collapse
Affiliation(s)
- Christian Witte
- University of Glasgow, Division of Biomedical Engineering, G12 8LT, Scotland
| | | | | | | | | | | | | |
Collapse
|
28
|
Mishra A, Kwon JS, Thakur R, Wereley S. Optoelectrical microfluidics as a promising tool in biology. Trends Biotechnol 2014; 32:414-21. [DOI: 10.1016/j.tibtech.2014.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 05/29/2014] [Accepted: 06/02/2014] [Indexed: 01/29/2023]
|
29
|
Fan Q, Hu W, Ohta AT. Laser-induced microbubble poration of localized single cells. LAB ON A CHIP 2014; 14:1572-8. [PMID: 24632785 PMCID: PMC4004443 DOI: 10.1039/c3lc51394g] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Laser-induced microbubbles were used to porate the cell membranes of localized single NIH/3T3 fibroblasts. Microsecond laser pulses were focused on an optically absorbent substrate, creating a vapour microbubble that oscillated in size at the laser focal point in a fluidic chamber. The shear stress accompanying the bubble size oscillation was able to porate nearby cells. Cell poration was demonstrated with the delivery of FITC-dextran dye with various molecular weights. Under optimal poration conditions, the cell poration efficiency was up to 95.2 ± 4.8%, while maintaining 97.6 ± 2.4% cell viability. The poration system is able to target a single cell without disturbing surrounding cells.
Collapse
Affiliation(s)
- Qihui Fan
- Department of Mechanical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 302, Honolulu, USA. Fax: +1-808-956-3427; Tel: 808-956-3427
| | - Wenqi Hu
- Department of Electrical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 483, Honolulu, USA
| | - Aaron T. Ohta
- Department of Electrical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 483, Honolulu, USA
| |
Collapse
|
30
|
Park S, Bassat DB, Yossifon G. Individually addressable multi-chamber electroporation platform with dielectrophoresis and alternating-current-electro-osmosis assisted cell positioning. BIOMICROFLUIDICS 2014; 8:024117. [PMID: 24803966 PMCID: PMC4000404 DOI: 10.1063/1.4873439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/16/2014] [Indexed: 05/09/2023]
Abstract
A multi-functional microfluidic platform was fabricated to demonstrate the feasibility of on-chip electroporation integrated with dielectrophoresis (DEP) and alternating-current-electro-osmosis (ACEO) assisted cell/particle manipulation. A spatial gradient of electroporation parameters was generated within a microchamber array and validated using normal human dermal fibroblast (NHDF) cells and red fluorescent protein-expressing human umbilical vein endothelial cells (RFP-HUVECs) with various fluorescent indicators. The edge of the bottom electrode, coinciding with the microchamber entrance, may act as an on-demand gate, functioning under either positive or negative DEP. In addition, at sufficiently low activation frequencies, ACEO vortices can complement the DEP to contribute to a rapid trapping/alignment of particles. As such, results clearly indicate that the microfluidic platform has the potential to achieve high-throughput screening for electroporation with spatial control and uniformity, assisted by DEP and ACEO manipulation/trapping of particles/cells into individual microchambers.
Collapse
Affiliation(s)
- Sinwook Park
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion-Israel Institute of Technology, Technion City 32000, Israel
| | - Dana Ben Bassat
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion-Israel Institute of Technology, Technion City 32000, Israel
| | - Gilad Yossifon
- Faculty of Mechanical Engineering, Micro- and Nanofluidics Laboratory, Technion-Israel Institute of Technology, Technion City 32000, Israel
| |
Collapse
|
31
|
Wang CH, Lee YH, Kuo HT, Liang WF, Li WJ, Lee GB. Dielectrophoretically-assisted electroporation using light-activated virtual microelectrodes for multiple DNA transfection. LAB ON A CHIP 2014; 14:592-601. [PMID: 24322338 DOI: 10.1039/c3lc51102b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Gene transfection is an important technology for various biological applications. The exogenous DNA is commonly delivered into cells by using a strong electrical field to form transient pores in cellular membranes. However, the high voltage required in this electroporation process may cause cell damage. In this study, a dielectrophoretically-assisted electroporation was developed by using light-activated virtual microelectrodes in a new microfluidic platform. The DNA electrotransfection used a low applied voltage and an alternating current to enable electroporation and transfection. Single or triple fluorescence-carrying plasmids were effectively transfected into various types of mammalian cells, and the fluorescent proteins were successfully expressed in live transfected cells. Moreover, the multi-triangle optical pattern that was projected onto a photoconductive layer to generate localized non-uniform virtual electric fields was found to have high transfection efficiency. The developed dielectrophoretically-assisted electroporation platform may provide a simpler system for gene transfection and could be widely applied in many biotechnological fields.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| | | | | | | | | | | |
Collapse
|
32
|
Dudani JS, Go DE, Gossett DR, Tan AP, Di Carlo D. Mediating Millisecond Reaction Time around Particles and Cells. Anal Chem 2014; 86:1502-10. [DOI: 10.1021/ac402920m] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Jaideep S. Dudani
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
| | - Derek E. Go
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
| | - Daniel R. Gossett
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
- California NanoSystems Institute, Los Angeles, California 90095, United States
| | - Andrew P. Tan
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
| | - Dino Di Carlo
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
- California NanoSystems Institute, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, Los
Angeles, California 90095, United States
| |
Collapse
|
33
|
Kremer C, Witte C, Neale SL, Reboud J, Barrett MP, Cooper JM. Shape-dependent optoelectronic cell lysis. Angew Chem Int Ed Engl 2014; 53:842-6. [PMID: 24402800 PMCID: PMC4441254 DOI: 10.1002/anie.201307751] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Indexed: 11/16/2022]
Abstract
We show an electrical method to break open living cells amongst a population of different cell types, where cell selection is based upon their shape. We implement the technique on an optoelectronic platform, where light, focused onto a semiconductor surface from a video projector creates a reconfigurable pattern of electrodes. One can choose the area of cells to be lysed in real-time, from single cells to large areas, simply by redrawing the projected pattern. We show that the method, based on the "electrical shadow" that the cell casts, allows the detection of rare cell types in blood (including sleeping sickness parasites), and has the potential to enable single cell studies for advanced molecular diagnostics, as well as wider applications in analytical chemistry.
Collapse
Affiliation(s)
- Clemens Kremer
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Rankine BuildingOakfield Avenue, Glasgow G12 8LT (UK)
| | - Christian Witte
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Rankine BuildingOakfield Avenue, Glasgow G12 8LT (UK)
| | - Steven L Neale
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Rankine BuildingOakfield Avenue, Glasgow G12 8LT (UK)
| | - Julien Reboud
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Rankine BuildingOakfield Avenue, Glasgow G12 8LT (UK)
| | - Michael P Barrett
- Institute of Infection, Immunity & Inflammation, School of Medical, Veterinary & Life Sciences, and Wellcome Centre for Molecular Parasitology, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building120 University Place, Glasgow G12 8TA (UK)
| | - Jonathan M Cooper
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Rankine BuildingOakfield Avenue, Glasgow G12 8LT (UK)
| |
Collapse
|
34
|
Kremer C, Witte C, Neale SL, Reboud J, Barrett MP, Cooper JM. Shape-Dependent Optoelectronic Cell Lysis. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201307751] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Abstract
Electroporation is a simple yet powerful technique for breaching the cell membrane barrier. The applications of electroporation can be generally divided into two categories: the release of intracellular proteins, nucleic acids and other metabolites for analysis and the delivery of exogenous reagents such as genes, drugs and nanoparticles with therapeutic purposes or for cellular manipulation. In this review, we go over the basic physics associated with cell electroporation and highlight recent technological advances on microfluidic platforms for conducting electroporation. Within the context of its working mechanism, we summarize the accumulated knowledge on how the parameters of electroporation affect its performance for various tasks. We discuss various strategies and designs for conducting electroporation at the microscale and then focus on analysis of intracellular contents and delivery of exogenous agents as two major applications of the technique. Finally, an outlook for future applications of microfluidic electroporation in increasingly diverse utilities is presented.
Collapse
Affiliation(s)
- Tao Geng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Chang Lu
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, USA. Fax: +1-540-231-5022; Tel: +1-540-231-8681
- School of Biomedical Engineering and Sciences, Virginia Tech-Wake Forest University, Blacksburg, VA 24061, USA
| |
Collapse
|
36
|
Yang SM, Tseng SY, Chen HP, Hsu L, Liu CH. Cell patterning via diffraction-induced optoelectronic dielectrophoresis force on an organic photoconductive chip. LAB ON A CHIP 2013; 13:3893-902. [PMID: 23925640 DOI: 10.1039/c3lc50351h] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
A laser diffraction-induced dielectrophoresis (DEP) phenomenon for the patterning and manipulation of individual HepG2 cells and polystyrene beads via positive/negative DEP forces is reported in this paper. The optoelectronic substrate was fabricated using an organic photoconductive material, TiOPc, via a spin-coating process on an indium tin oxide glass surface. A piece of square aperture array grid grating was utilized to transform the collimating He-Ne laser beam into the multi-spot diffraction pattern which forms the virtual electrodes as the TiOPc-coating surface was illuminated by the multi-spot diffraction light pattern. HepG2 cells were trapped at the spot centers and polystyrene beads were trapped within the dim region of the illuminated image. The simulation results of light-induced electric field and a Fresnel diffraction image illustrated the distribution of trapped microparticles. The HepG2 morphology change, adhesion, and growth during a 5-day culture period demonstrated the cell viability through our manipulation. The power density inducing DEP phenomena, the characteristics of the thin TiOPc coating layer, the operating ac voltage/frequency, the sandwiched medium, the temperature rise due to the ac electric fields and the illuminating patterns are discussed in this paper. This concept of utilizing laser diffraction images to generate virtual electrodes on our TiOPc-based optoelectronic DEP chip extends the applications of optoelectronic dielectrophoretic manipulation.
Collapse
Affiliation(s)
- Shih-Mo Yang
- Department of Mechanical and Automation Engineering, Chinese University of Hong Kong, Hong Kong
| | | | | | | | | |
Collapse
|
37
|
Huang KW, Wu YC, Lee JA, Chiou PY. Microfluidic integrated optoelectronic tweezers for single-cell preparation and analysis. LAB ON A CHIP 2013; 13:3721-7. [PMID: 23884358 DOI: 10.1039/c3lc50607j] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
We report a novel microfluidic integrated optoelectronic tweezers (OET) platform for single-cell sample preparation and analysis. Integration of OET and microfluidics is achieved by embedding single-wall carbon nanotube (SWNT) electrodes into multilayer PDMS structures. This integrated platform allows users to selectively pick up individual cells from a population with light beams based on their optical signatures such as size, shape, and fluorescence, and transport them into isolated chambers using light induced dielectrophoretic forces. Isolated cells can be encapsulated into nanoliter liquid plugs and transported out of the platform for downstream molecule analysis using standard commercial instruments.
Collapse
Affiliation(s)
- Kuo-Wei Huang
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
38
|
|
39
|
Huang KW, Su TW, Ozcan A, Chiou PY. Optoelectronic tweezers integrated with lensfree holographic microscopy for wide-field interactive cell and particle manipulation on a chip. LAB ON A CHIP 2013; 13:2278-84. [PMID: 23661233 PMCID: PMC3684708 DOI: 10.1039/c3lc50168j] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We demonstrate an optoelectronic tweezer (OET) coupled to a lensfree holographic microscope for real-time interactive manipulation of cells and micro-particles over a large field-of-view (FOV). This integrated platform can record the holographic images of cells and particles over the entire active area of a CCD sensor array, perform digital image reconstruction to identify target cells, dynamically track the positions of cells and particles, and project light beams to trigger light-induced dielectrophoretic forces to pattern and sort cells on a chip. OET technology has been previously shown to be capable of performing parallel single cell manipulation over a large area. However, its throughput has been bottlenecked by the number of cells that can be imaged within the limited FOV of a conventional microscope objective lens. Integrating lensfree holographic imaging with OET solves this fundamental FOV barrier, while also creating a compact on-chip cell/particle manipulation platform. Using this unique platform, we have successfully demonstrated real-time interactive manipulation of thousands of single cells and micro-particles over an ultra-large area of e.g., 240 mm(2) (i.e. 17.96 mm × 13.52 mm).
Collapse
Affiliation(s)
- Kuo-Wei Huang
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
40
|
Longsine-Parker W, Wang H, Koo C, Kim J, Kim B, Jayaraman A, Han A. Microfluidic electro-sonoporation: a multi-modal cell poration methodology through simultaneous application of electric field and ultrasonic wave. LAB ON A CHIP 2013; 13:2144-52. [PMID: 23615834 DOI: 10.1039/c3lc40877a] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
A microfluidic device that simultaneously applies the conditions required for microelectroporation and microsonoporation in a flow-through scheme toward high-efficiency and high-throughput molecular delivery into mammalian cells is presented. This multi-modal poration microdevice using simultaneous application of electric field and ultrasonic wave was realized by a three-dimensional (3D) microelectrode scheme where the electrodes function as both electroporation electrodes and cell flow channel so that acoustic wave can be applied perpendicular to the electric field simultaneously to cells flowing through the microfluidic channel. This 3D microelectrode configuration also allows a uniform electric field to be applied while making the device compatible with fluorescent microscopy. It is hypothesized that the simultaneous application of two different fields (electric field and acoustic wave) in perpendicular directions allows formation of transient pores along two axes of the cell membrane at reduced poration intensities, hence maximizing the delivery efficiency while minimizing cell death. The microfluidic electro-sonoporation system was characterized by delivering small molecules into mammalian cells, and showed average poration efficiency of 95.6% and cell viability of 97.3%. This proof of concept result shows that by combining electroporation and sonoporation together, significant improvement in molecule delivery efficiency could be achieved while maintaining high cell viability compared to electroporation or sonoporation alone. The microfluidic electro-sonoporation device presented here is, to the best of our knowledge, the first multi-modal cell poration device using simultaneous application of electric field and ultrasonic wave. This new multi-modal cell poration strategy and system is expected to have broad applications in delivery of small molecule therapeutics and ultimately in large molecule delivery such as gene transfection applications where high delivery efficiency and high viability are crucial.
Collapse
Affiliation(s)
- Whitney Longsine-Parker
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Li ZG, Liu AQ, Klaseboer E, Zhang JB, Ohl CD. Single cell membrane poration by bubble-induced microjets in a microfluidic chip. LAB ON A CHIP 2013; 13:1144-50. [PMID: 23364762 DOI: 10.1039/c3lc41252k] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
This paper demonstrates membrane poration of a single suspension cell due to a fast liquid microjet. The jet is formed during the collapse of a laser induced bubble created at a variable stand-off distance from the target cell. The cell is trapped by a converging structure within a microfluidic chip. The asymmetrical growth and collapse of the cavitation bubble next to the cell lead to the microjetting, which deforms and porates the cell membrane. In the experiments, the membrane porations of myeloma cells are probed with the uptake of trypan blue. Time-resolved studies of the diffusion of trypan blue show a marked dependency on the bubble dynamics, i.e. the stand-off distance. The penetration length of the dye increases with shorter distances. Numerical simulations of the diffusion process agree with larger pores formed on the cell membrane. This method allows for a fast, repeatable, and localized rupture of membranes of individual cells in suspension.
Collapse
Affiliation(s)
- Z G Li
- Division of Microelectronics, School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | | | | | | | | |
Collapse
|
42
|
Hung SH, Huang SC, Lee GB. Numerical simulation of optically-induced dielectrophoresis using a voltage-transformation-ratio model. SENSORS 2013; 13:1965-83. [PMID: 23385411 PMCID: PMC3649436 DOI: 10.3390/s130201965] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 01/16/2013] [Accepted: 01/29/2013] [Indexed: 11/16/2022]
Abstract
Optically-induced dielectrophoresis (ODEP) has been extensively used for the manipulation and separation of cells, beads and micro-droplets in microfluidic devices. With this approach, non-uniform electric fields induced by light projected on a photoconductive layer can be used to generate attractive or repulsive forces on dielectric materials. Then, moving these light patterns can be used for the manipulation of particles in the microfluidic devices. This study reports on the results from numerical simulation of the ODEP platform using a new model based on a voltage transformation ratio, which takes the effective electrical voltage into consideration. Results showed that the numerical simulation was in reasonably agreement with experimental data for the manipulation of polystyrene beads and emulsion droplets, with a coefficient of variation less than 6.2% (n = 3). The proposed model can be applied to simulations of the ODEP force and may provide a reliable tool for estimating induced dielectrophoretic forces and electric fields, which is crucial for microfluidic applications.
Collapse
Affiliation(s)
- Shih-Hsun Hung
- Department of Engineering Science, National Cheng Kung University, Tainan 701, Taiwan; E-Mail:
| | - Sheng-Chieh Huang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mail:
| | - Gwo-Bin Lee
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-3-571-5131 (ext. 33765); Fax: +886-3-574-2495
| |
Collapse
|
43
|
Hai A, Spira ME. On-chip electroporation, membrane repair dynamics and transient in-cell recordings by arrays of gold mushroom-shaped microelectrodes. LAB ON A CHIP 2012; 12:2865-73. [PMID: 22678065 DOI: 10.1039/c2lc40091j] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
This study demonstrates the use of on-chip gold mushroom-shaped microelectrodes (gMμEs) to generate localized electropores in the plasma membrane of adhering cultured neurons and to electrophysiologically monitor the ensuing membrane repair dynamics. Delivery of an alternating voltage pulse (0.5-1 V, 100 Hz, 300 ms) through an extracellularly positioned micrometer-sized gMμE electroporates the patch of plasma membrane facing the microelectrode. The repair dynamics of the electropores were analyzed by continuous monitoring of the neuron transmembrane potential, input resistance (R(in)) and action potential (AP) amplitude with an intracellular microelectrode and a number of neighbouring extracellular gMμEs. Electroporation by a gMμE is associated with local elevation of the free intracellular calcium concentration ([Ca(2+)](i)) around the gMμE. The membrane repair kinetics proceeds as an exponential process interrupted by abrupt recovery steps. These abrupt events are consistent with the "membrane patch model" of membrane repair in which patches of intracellular membrane fuse with the plasma membrane at the site of injury. Membrane electroporation by a single gMμE generates a neuron-gMμE configuration that permits recordings of attenuated intracellular action potentials. We conclude that the use of on-chip cultured neurons via a gMμE configuration provides a unique neuroelectronic interface that enables the selection of individual cells for electroporation, generates a confined electroporated membrane patch, monitors membrane repair dynamics and records attenuated intracellular action potentials.
Collapse
Affiliation(s)
- Aviad Hai
- Department of Neurobiology the Life Sciences Institute, and the Harvey M. Kruger Family center for Nanoscience and Nanotechnology. The Hebrew University of Jerusalem, Jerusalem, Israel
| | | |
Collapse
|
44
|
Bridle H, Kersaudy-Kerhoas M, Miller B, Gavriilidou D, Katzer F, Innes EA, Desmulliez MPY. Detection of Cryptosporidium in miniaturised fluidic devices. WATER RESEARCH 2012; 46:1641-1661. [PMID: 22305660 DOI: 10.1016/j.watres.2012.01.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 01/11/2012] [Accepted: 01/12/2012] [Indexed: 05/28/2023]
Abstract
Contamination of drinking water with the protozoan pathogen, Cryptosporidium, represents a serious risk to human health due to the low infectious dose and the resistance of this parasite to chlorine disinfection. Therefore, several countries have legislated for the frequent monitoring of drinking water for Cryptosporidium presence. Existing approved monitoring protocols are however time-consuming and do not provide essential information on the species, virulence or viability of detected oocysts. Rapid, more information-rich and automatable systems for Cryptosporidium detection are highly sought-after, and numerous miniaturised devices have been developed to address this need. This review article aims to summarise the state-of-the-art and compare the performance of these systems in terms of detection limit, ability to determine species, viability and performance in the presence of interferents. Finally, conclusions are drawn with regard to the most promising methods and directions of future research.
Collapse
Affiliation(s)
- Helen Bridle
- University of Edinburgh, King's Buildings, Edinburgh, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
45
|
Low-frequency ac electroporation shows strong frequency dependence and yields comparable transfection results to dc electroporation. J Control Release 2012; 160:570-6. [PMID: 22516092 DOI: 10.1016/j.jconrel.2012.04.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 04/01/2012] [Indexed: 01/01/2023]
Abstract
Conventional electroporation has been conducted by employing short direct current (dc) pulses for delivery of macromolecules such as DNA into cells. The use of alternating current (ac) field for electroporation has mostly been explored in the frequency range of 10kHz-1MHz. Based on Schwan equation, it was thought that with low ac frequencies (10Hz-10kHz), the transmembrane potential does not vary with the frequency. In this report, we utilized a flow-through electroporation technique that employed continuous 10Hz-10kHz ac field (based on either sine waves or square waves) for electroporation of cells with defined duration and intensity. Our results reveal that electropermeabilization becomes weaker with increased frequency in this range. In contrast, transfection efficiency with DNA reaches its maximum at medium frequencies (100-1000Hz) in the range. We postulate that the relationship between the transfection efficiency and the ac frequency is determined by combined effects from electrophoretic movement of DNA in the ac field, dependence of the DNA/membrane interaction on the ac frequency, and variation of transfection under different electropermeabilization intensities. The fact that ac electroporation in this frequency range yields high efficiency for transfection (up to ~71% for Chinese hamster ovary cells) and permeabilization suggests its potential for gene delivery.
Collapse
|
46
|
Kim J, Hwang I, Britain D, Chung TD, Sun Y, Kim DH. Microfluidic approaches for gene delivery and gene therapy. LAB ON A CHIP 2011; 11:3941-8. [PMID: 22027752 DOI: 10.1039/c1lc20766k] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Recent advances in microfluidics have created new and exciting prospects for gene delivery and therapy. The micro-scaled environment within microfluidic systems enables precise control and optimization of multiple processes and techniques used in gene transfection and the production of gene and drug transporters. Traditional non-viral gene transfection methods, such as electroporation, microinjection and optical gene transfection, are improved from the use of innovative microfluidic systems. Additionally, microfluidic systems have also made the production of many viral and non-viral vectors controlled, automated, and reproducible. In summary, the development and application of microfluidic systems are producing increased efficiency in gene delivery and promise improved gene therapy results.
Collapse
Affiliation(s)
- Jungkyu Kim
- Department of Chemistry and Electrical Engineering, University of California, Berkeley, CA, USA
| | | | | | | | | | | |
Collapse
|
47
|
Xu Y, Yao H, Wang L, Xing W, Cheng J. The construction of an individually addressable cell array for selective patterning and electroporation. LAB ON A CHIP 2011; 11:2417-23. [PMID: 21625729 DOI: 10.1039/c1lc20183b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
In basic cell biology research and drug discovery, it is important to rapidly introduce genes, proteins or drug compounds into cells without permanent damage. Here, we report a three dimensional SU-8 micro-well structure sandwiched with an indium tin oxide (ITO) electrode-covered slide from the top and an individually addressable array of microelectrodes on the bottom to allow parallel delivery of exogenous molecules into various cells in a spatially specific manner. A positive dielectrophoretic force was selectively applied by energizing appropriate electrodes to capture the dispersed cells at the bottom electrode, while the micro-wells were designed to confine cells in situ when the positive dielectrophoretic force is removed. The combination of spatial positive dielectrophoresis (pDEP) and micro-wells made it possible to construct cell microarrays with specific patterns. Once the cells become attached to the electrodes, different plasmids can be introduced sequentially for selective electroporation. The present cell arraying-assisted electroporation chip integrates a pDEP-assisted cell positioning function with selective electroporation to provide a simple and efficient method for gene transfer. This platform is ideal for high throughput screening of compounds in parallel and thus holds promise for applications in cellular and molecular research.
Collapse
Affiliation(s)
- Youchun Xu
- Medical Systems Biology Research Center, Tsinghua University, Beijing, China
| | | | | | | | | |
Collapse
|
48
|
Kumar A, Williams SJ, Chuang HS, Green NG, Wereley ST. Hybrid opto-electric manipulation in microfluidics-opportunities and challenges. LAB ON A CHIP 2011; 11:2135-48. [PMID: 21603691 DOI: 10.1039/c1lc20208a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Hybrid opto-electric manipulation in microfluidics/nanofluidics refers to a set of methodologies employing optical modulation of electrokinetic schemes to achieve particle or fluid manipulation at the micro- and nano-scale. Over the last decade, a set of methodologies, which differ in their modulation strategy and/or the length scale of operation, have emerged. These techniques offer new opportunities with their dynamic nature, and their ability for parallel operation has created novel applications and devices. Hybrid opto-electric techniques have been utilized to manipulate objects ranging in diversity from millimetre-sized droplets to nano-particles. This review article discusses the underlying principles, applications and future perspectives of various hybrid opto-electric techniques that have emerged over the last decade under a unified umbrella.
Collapse
Affiliation(s)
- Aloke Kumar
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, USA.
| | | | | | | | | |
Collapse
|
49
|
Valley JK, Pei SN, Jamshidi A, Hsu HY, Wu MC. A unified platform for optoelectrowetting and optoelectronic tweezers. LAB ON A CHIP 2011; 11:1292-7. [PMID: 21311817 DOI: 10.1039/c0lc00568a] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A platform capable of seamlessly unifying both optoelectrowetting and optoelectronic tweezers is presented. This enables the user to manipulate aqueous droplets (with electrowetting) as well as individual particles within those droplets (with dielectrophoresis). The device requires no photolithography and droplet/particle manipulation can occur continuously over the entire surface of the device. Droplet and 10 µm polystyrene particle speeds of up to 8 mm s(-1) and 60 µm s(-1), respectively, are demonstrated. Particle concentration within, and subsequent splitting of, a droplet is performed resulting in average concentration efficiencies of 93%. Serial concentration is also demonstrated resulting in exponentially increasing particle concentrations and a 10× concentration increase. Finally, the platform is used to select a single cell out of a cohort and subsequently encapsulate it in its own aqueous droplet.
Collapse
Affiliation(s)
- Justin K Valley
- Berkeley Sensor and Actuator Center, Department of Electrical Engineering and Computer Science, University of California Berkeley, 497 Cory Hall, Berkeley, CA 94720, USA.
| | | | | | | | | |
Collapse
|
50
|
Zarowna-Dabrowska A, Neale SL, Massoubre D, McKendry J, Rae BR, Henderson RK, Rose MJ, Yin H, Cooper JM, Gu E, Dawson MD. Miniaturized optoelectronic tweezers controlled by GaN micro-pixel light emitting diode arrays. OPTICS EXPRESS 2011; 19:2720-8. [PMID: 21369093 DOI: 10.1364/oe.19.002720] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
A novel, miniaturized optoelectronic tweezers (OET) system has been developed using a CMOS-controlled GaN micro-pixelated light emitting diode (LED) array as an integrated micro-light source. The micro-LED array offers spatio-temporal and intensity control of the emission pattern, enabling the creation of reconfigurable virtual electrodes to achieve OET. In order to analyse the mechanism responsible for particle manipulation in this OET system, the average particle velocity, electrical field and forces applied to the particles were characterized and simulated. The capability of this miniaturized OET system for manipulating and trapping multiple particles including polystyrene beads and live cells has been successfully demonstrated.
Collapse
|